Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

BET protein Brd4 activates transcription in neurons and BET inhibitor Jq1 blocks memory in mice

Abstract

Precise regulation of transcription is crucial for the cellular mechanisms underlying memory formation. However, the link between neuronal stimulation and the proteins that directly interact with histone modifications to activate transcription in neurons remains unclear. Brd4 is a member of the bromodomain and extra-terminal domain (BET) protein family, which binds acetylated histones and is a critical regulator of transcription in many cell types, including transcription in response to external cues. Small molecule BET inhibitors are in clinical trials, yet almost nothing is known about Brd4 function in the brain. Here we show that Brd4 mediates the transcriptional regulation underlying learning and memory. The loss of Brd4 function affects critical synaptic proteins, which results in memory deficits in mice but also decreases seizure susceptibility. Thus Brd4 provides a critical link between neuronal activation and the transcriptional responses that occur during memory formation.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Brd4 is expressed in neurons throughout the brain.
Figure 2: Brd4 regulates IEG transcription in response to stimulation in neurons.
Figure 3: Genome-wide analysis of effects of Jq1.
Figure 4: CK2 regulates Brd4 in neurons.
Figure 5: Phosphorylation of Brd4 is critical for its function.
Figure 6: Brd4 regulates synaptic receptor subunit GluA1.
Figure 7: Jq1 affects mouse behavior.
Figure 8: Jq1 decreases seizure susceptibility.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Hsieh, J. & Gage, F.H. Chromatin remodeling in neural development and plasticity. Curr. Opin. Cell Biol. 17, 664–671 (2005).

    CAS  PubMed  Google Scholar 

  2. Gapp, K., Woldemichael, B.T., Bohacek, J. & Mansuy, I.M. Epigenetic regulation in neurodevelopment and neurodegenerative diseases. Neuroscience 264, 99–111 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Frey, U., Frey, S., Schollmeier, F. & Krug, M. Influence of actinomycin D, a RNA synthesis inhibitor, on long-term potentiation in rat hippocampal neurons in vivo and in vitro. J. Physiol. (Lond.) 490, 703–711 (1996).

    Article  CAS  Google Scholar 

  4. Frey, U., Krug, M., Brödemann, R., Reymann, K. & Matthies, H. Long-term potentiation induced in dendrites separated from rat's CA1 pyramidal somata does not establish a late phase. Neurosci. Lett. 97, 135–139 (1989).

    Article  CAS  PubMed  Google Scholar 

  5. Nguyen, P.V., Abel, T. & Kandel, E.R. Requirement of a critical period of transcription for induction of a late phase of LTP. Science 265, 1104–1107 (1994).

    Article  CAS  PubMed  Google Scholar 

  6. Messaoudi, E., Ying, S.-W., Kanhema, T., Croll, S.D. & Bramham, C.R. Brain-derived neurotrophic factor triggers transcription-dependent, late phase long-term potentiation in vivo. J. Neurosci. 22, 7453–7461 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Sims, R.J., Belotserkovskaya, R. & Reinberg, D. Elongation by RNA polymerase II: the short and long of it. Genes Dev. 18, 2437–2468 (2004).

    Article  CAS  PubMed  Google Scholar 

  8. Kadonaga, J.T. Regulation of RNA polymerase II transcription by sequence-specific DNA binding factors. Cell 116, 247–257 (2004).

    Article  CAS  PubMed  Google Scholar 

  9. Jang, M.K. et al. The bromodomain protein Brd4 is a positive regulatory component of P-TEFb and stimulates RNA polymerase II-dependent transcription. Mol. Cell 19, 523–534 (2005).

    Article  CAS  PubMed  Google Scholar 

  10. Liu, W. et al. Brd4 and JMJD6-associated anti-pause enhancers in regulation of transcriptional pause release. Cell 155, 1581–1595 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zippo, A. et al. Histone crosstalk between H3S10ph and H4K16ac generates a histone code that mediates transcription elongation. Cell 138, 1122–1136 (2009).

    Article  CAS  PubMed  Google Scholar 

  12. Nagarajan, S. et al. Bromodomain protein BRD4 is required for estrogen receptor-dependent enhancer activation and gene transcription. Cell Rep. 8, 460–469 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Belkina, A.C. & Denis, G.V. BET domain co-regulators in obesity, inflammation and cancer. Nat. Rev. Cancer 12, 465–477 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Wu, S.-Y., Lee, A.-Y., Lai, H.-T., Zhang, H. & Chiang, C.-M. Phospho switch triggers Brd4 chromatin binding and activator recruitment for gene-specific targeting. Mol. Cell 49, 843–857 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Houzelstein, D. et al. Growth and early postimplantation defects in mice deficient for the bromodomain-containing protein Brd4. Mol. Cell. Biol. 22, 3794–3802 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Descalzi, G., Fukushima, H., Suzuki, A., Kida, S. & Zhuo, M. Genetic enhancement of neuropathic and inflammatory pain by forebrain upregulation of CREB-mediated transcription. Mol. Pain 8, 90 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zuber, J. et al. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature 478, 524–528 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Filippakopoulos, P. et al. Selective inhibition of BET bromodomains. Nature 468, 1067–1073 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hargreaves, D.C., Horng, T. & Medzhitov, R. Control of inducible gene expression by signal-dependent transcriptional elongation. Cell 138, 129–145 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Saha, R.N. et al. Rapid activity-induced transcription of Arc and other IEGs relies on poised RNA polymerase II. Nat. Neurosci. 14, 848–856 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Fischer, A., Sananbenesi, F., Mungenast, A. & Tsai, L.-H. Targeting the correct HDAC(s) to treat cognitive disorders. Trends Pharmacol. Sci. 31, 605–617 (2010).

    Article  CAS  PubMed  Google Scholar 

  22. Schael, S. et al. Casein kinase 2 phosphorylation of protein kinase C and casein kinase 2 substrate in neurons (PACSIN) 1 protein regulates neuronal spine formation. J. Biol. Chem. 288, 9303–9312 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Delmore, J.E. et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146, 904–917 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Mirguet, O. et al. Discovery of epigenetic regulator I-BET762: lead optimization to afford a clinical candidate inhibitor of the BET bromodomains. J. Med. Chem. 56, 7501–7515 (2013).

    CAS  PubMed  Google Scholar 

  25. Asangani, I.A. et al. Therapeutic targeting of BET bromodomain proteins in castration-resistant prostate cancer. Nature 510, 278–282 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kanno, T. et al. BRD4 assists elongation of both coding and enhancer RNAs by interacting with acetylated histones. Nat. Struct. Mol. Biol. 21, 1047–1057 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Lussier, M.P., Gu, X., Lu, W. & Roche, K.W. Casein kinase 2 phosphorylates GluA1 and regulates its surface expression. Eur. J. Neurosci. 39, 1148–1158 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Sanz-Clemente, A., Matta, J.A., Isaac, J.T.R. & Roche, K.W. Casein kinase 2 regulates the NR2 subunit composition of synaptic NMDA receptors. Neuron 67, 984–996 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Dietrich, J.-B. The MEF2 family and the brain: from molecules to memory. Cell Tissue Res. 352, 179–190 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Korb, E. & Finkbeiner, S. Arc in synaptic plasticity: from gene to behavior. Trends Neurosci. 34, 591–598 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Borges, K. & Dingledine, R. Functional organization of the GluR1 glutamate receptor promoter. J. Biol. Chem. 276, 25929–25938 (2001).

    Article  CAS  PubMed  Google Scholar 

  32. Myers, S.J. et al. Transcriptional regulation of the GluR2 gene: neural-specific expression, multiple promoters, and regulatory elements. J. Neurosci. 18, 6723–6739 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Matzuk, M.M. et al. Small-molecule inhibition of BRDT for male contraception. Cell 150, 673–684 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Antunes, M. & Biala, G. The novel object recognition memory: neurobiology, test procedure, and its modifications. Cogn. Process. 13, 93–110 (2012).

    Article  CAS  PubMed  Google Scholar 

  35. Naydenov, A.V. et al. ABHD6 blockade exerts antiepileptic activity in PTZ-induced seizures and in spontaneous seizures in R6/2 mice. Neuron 83, 361–371 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ferraro, T.N. et al. Mapping loci for pentylenetetrazol-induced seizure susceptibility in mice. J. Neurosci. 19, 6733–6739 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Dhir, A. Pentylenetetrazol (PTZ) kindling model of epilepsy. Curr. Protoc. Neurosci. Ch. 9, unit 9.37 (2012).

  38. Bialer, M. & White, H.S. Key factors in the discovery and development of new antiepileptic drugs. Nat. Rev. Drug Discov. 9, 68–82 (2010).

    Article  CAS  PubMed  Google Scholar 

  39. Brehme, H., Kirschstein, T., Schulz, R. & Köhling, R. In vivo treatment with the casein kinase 2 inhibitor 4,5,6,7- tetrabromotriazole augments the slow afterhyperpolarizing potential and prevents acute epileptiform activity. Epilepsia 55, 175–183 (2014).

    Article  CAS  PubMed  Google Scholar 

  40. Rogawski, M.A. AMPA receptors as a molecular target in epilepsy therapy. Acta Neurol. Scand. Suppl. 2013, 9–18, (2013).

    Article  Google Scholar 

  41. Zhang, J. & Abdullah, J.M. The role of GluA1 in central nervous system disorders. Rev. Neurosci. 24, 499–505 (2013).

    Article  CAS  PubMed  Google Scholar 

  42. Kato, A.S., Gill, M.B., Yu, H., Nisenbaum, E.S. & Bredt, D.S. TARPs differentially decorate AMPA receptors to specify neuropharmacology. Trends Neurosci. 33, 241–248 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Yamaguchi, S., Donevan, S.D. & Rogawski, M.A. Anticonvulsant activity of AMPA/kainate antagonists: comparison of GYKI 52466 and NBOX in maximal electroshock and chemoconvulsant seizure models. Epilepsy Res. 15, 179–184 (1993).

    Article  CAS  PubMed  Google Scholar 

  44. Namba, T., Morimoto, K., Sato, K., Yamada, N. & Kuroda, S. Antiepileptogenic and anticonvulsant effects of NBQX, a selective AMPA receptor antagonist, in the rat kindling model of epilepsy. Brain Res. 638, 36–44 (1994).

    Article  CAS  PubMed  Google Scholar 

  45. Hara, H. et al. Effect of YM872, a selective and highly water-soluble AMPA receptor antagonist, in the rat kindling and rekindling model of epilepsy. Eur. J. Pharmacol. 531, 59–65 (2006).

    Article  CAS  PubMed  Google Scholar 

  46. Kodama, M. et al. Effects of YM90K, a selective AMPA receptor antagonist, on amygdala-kindling and long-term hippocampal potentiation in the rat. Eur. J. Pharmacol. 374, 11–19 (1999).

    Article  CAS  PubMed  Google Scholar 

  47. Mattes, H., Carcache, D., Kalkman, H.O. & Koller, M. α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) antagonists: from bench to bedside. J. Med. Chem. 53, 5367–5382 (2010).

    Article  CAS  PubMed  Google Scholar 

  48. Rakhade, S.N. et al. Glutamate receptor 1 phosphorylation at serine 831 and 845 modulates seizure susceptibility and hippocampal hyperexcitability after early life seizures. J. Neurosci. 32, 17800–17812 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Banaszynski, L.A. et al. Hira-dependent histone H3.3 deposition facilitates PRC2 recruitment at developmental loci in ES cells. Cell 155, 107–120 (2013).

    Article  CAS  PubMed  Google Scholar 

  50. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Trapnell, C. et al. Differential analysis of gene regulation at transcript resolution with RNA-seq. Nat. Biotechnol. 31, 46–53 (2013).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank K. Ozato for the Brd4 construct, J. Gresack and the Rockefeller behavioral core for guidance and suggestions, K. Thomas for microscopy expertise, the Memorial Sloan-Kettering Cancer Center histology core for immunohistochemistry support, A. Soshnev for the schematic model graphic, J. Gerace for editing and members of the Allis laboratory for feedback. Support for this work was provided by a grant from the Robertson Foundation, a Ruth Kirschstein US National Research Service Award fellowship (F32MH103921), the US National Institutes of Health (R01 NS34389, NS081706) and a Simons Foundation Research Award. R.B.D. is an Investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

E.K. wrote the manuscript and designed and carried out experiments. M.H. helped carry out behavioral testing and molecular studies. I.Z.-S. helped with seizure testing. R.B.D. helped initiate and design the project and provided feedback, and C.D.A. provided support, feedback and guidance.

Corresponding authors

Correspondence to Erica Korb or C David Allis.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Brd4 is expressed in the brain.

(a) Whole cell lysate of regions of adult mouse brain. Representative blot of 3 biological replicates. (b, c) Cultured cortical neurons stained with Brd4 and CamKII (b) or Gaba (c). (d) Brd4 mRNA from cultured neurons after BDNF treatment (one sample t-test for 0.5 hours n = 14, for 2 hours n = 13, for 4 hours n = 14, for 8 hours n = 12, for 24 hours n = 13, P = 0.0339, t = 2.01). (e, f) Brd4 protein after short (e) or long (f) BDNF treatment. Representative blot of 3 biological replicates. *, p<0.05. Full-length blots are presented in Supplementary Figure 10. min, minutes. h, hours. Error bars represent standard error. Scale bar is 10 μm.

Supplementary Figure 2 Brd4 regulates IEG transcription in response neurons.

(a, b, c) Time course of the BDNF-induced increase in Arc (a), Fos (b) and Nr4a1 (c) after (-) or (+)Jq1 treatment. (d) Brd4 mRNA 3 days after lentiviral infection of an siRNA targeted against Brd4 compared to a scrambled siRNA (one-sample t test, n = 7 biological replicates P = 6.18E-6, t = 13.07). (e) BDNF-induced MAPK phosphorylation after Brd4 knockdown with lentiviral infection. Representative of 2 biological replicates. (f, g) Staining (f) and quantification (g) of Brd4 in neurons 5 days after transfection with GFP and Brd4 siRNA (unpaired two-sided t test n = 45 neurons for control siRNA and 37 neurons for Brd4 siRNA from 5 biological replicates, P = 6.38E-16, t = 10.093). (h, i) Brd2 (h) and Brd3 (i) mRNA fold decrease 5 days after transfection in N2A cells (one sample t-test n = 3 biological replicates, for Brd2 P = 0.0157, t = 5.51, for Brd3 P = 0.019, t = 4.92). (i) Quantification of Brd4 staining 5 days after transfection with different siRNAs targeted against Brd4. N = 47 neurons for control siRNA (upaired two-tailed t test for control n = 47 for Brd4 siRNA1 n = 43 and for Brd4 siRNA2 n = 37 neurons from 2 biological replicates, for siRNA1 P = 2.24E-38, t = 22.91, for siRNA2 P = 2.82E-25, t = 15.18). (k, l) Staining (k) and quantification (l) of Arc with or without 30 minutes BDNF stimulation after transfection with GFP and either a nontargeting siRNA or different Brd4 siRNA constructs (upaired two-tailed t test for control n = 25, for control + BDNF n = 23, for Brd4 siRNA1 n = 22, for Brd4 siRNA1 + BDNF n = 22, for Brd4 siRNA2 n = 14, and for Brd4 siRNA2 + BDNF n = 26 from 2 biological replicates, for control vs BDNF P = 9.53E-4, t = 3.432). (m) Quantification of Arc staining in neurons transfected with a long or short form of Brd4 (upaired two-tailed t test for control n = 42 neurons, for long Brd4 n = 26 neurons and for short Brd4 n = 30 neurons, for control vs long Brd4 P = 0.015, t = 2.48, for long vs short Brd4 P = 0.0016, t = 3.29). (n) Decrease in Arc, Fos and Nr4a1 RNA after 24 hours of Jq1 treatment in the absence of exogenous stimulation in cultured cortical neurons (one sample t test, for Arc n = 13 biological replicates P = 3.098E-5, t = 6.58, for Fos n = 13 biological replicates P = 0.013, t = 2.65, and for Nr4a1 n = 11, P = 0.00104, t = 4.75). (o) Jq1 does not affect global H3 or H4 acetyl levels. (p) MAPK phosphorylation in response to BDNF with or without Jq1 pretreatment. Representative blot of 2 biological replicates. a.u., arbitrary units. #, p< 0.05 for univariate analysis. ###, p< 0.001 for univariate analysis. *, p< 0.05. ***, p<0.001. Full-length blots are presented in Supplementary Figure 10. Error bars represent standard error. Scale bar is 10 μM.

Supplementary Figure 3 RNA-sequencing analysis of effects of Jq1 in neurons.

(a) Experimental paradigm for genome-wide analysis of effects of Jq1. (b) Heat map of fold induction of all genes significantly induced by BDNF after treatment with (-) or (+) Jq1. (c) Top GO terms of genes clusters enriched in Jq1 up-regulated genes. Data represents averages of 3 biological replicates. (d) Histone acetylation at promoter regions of IEGs in response to BDNF for acetyl marks that recruit Brd4. N = 3 biological replicates. (e) Western blot analysis of acetylated histone H4K16 and H3K14 with (-) or (+) Jq1. Representative blot of 3 biological replicates. Full-length blots are presented in Supplementary Figure 11. min, minutes.

Supplementary Figure 4 CK2 regulates Brd4 in neurons.

(a, b, c) Brd4 ChIP-qPCR analysis in neurons stimulated with BDNF to measure Brd4 at promoters of Arc (a), Fos (b), and Nr4a1 (c). (d, e, f) Brd2 ChIP in neurons stimulated with BDNF at promoters of Arc (d), Fos (e), and Nr4a1 (f). N = 3 biological replicates. (g, h, i) Brd3 ChIP in neurons stimulated with BDNF at promoters of Arc (g), Fos (h), and Nr4a1 (i). N = 3 biological replicates for a-i. (j) MAPK phosphorylation in response to BDNF in the presence of TBB. Representative of 3 biological replicates. (k, l, m) CBP ChIP-qPCR analysis in neurons stimulated with BDNF with Jq1 or TBB pretreatment to measure Brd4 at promoters of Arc (k), Fos (l), and Nr4a1 (m). N = 2 biological replicates. (n) Quantification of Ck2 expression in neurons transfected with GFP and either nontargeting siRNA, or siRNA targeting CK2 (unpaired two-tailed t test, for control siRNA n = 24 neurons and for CK2 siRNA n = 26 neurons from 3 biological replicates, P = 8.05E-5, t = 3.29). (o, p) Arc and CK2 staining (o) and Arc quantification (p) after a 30 minute BDNF stimulation of neurons transfected with GFP and either a nontargeting siRNA pool or a siRNA pool targeting CK2 (unpaired two-tailed t test, for control siRNA n = 50 neurons, n = 39 for BDNF, for CK2 siRNA n = 39, and for CK2 siRNA + BDNF n = 37 from 5 biological replicates, control vs BDNF, P = 1.21E-5, t = 5.244, for BDNF vs CK2 siRNA + BDNF P = 7.09E-7, t = 5.42). (q) Mobile fraction quantification of EGFP-Brd4 (unpaired two-tailed t test for control n = 15 for BDNF 18 neurons. P = 5.389E-5, t = 4.73). ***, p<0.001. a.u. arbitrary units. a.u. arbitrary units. min, minutes. Error bars represent standard error. Full-length blots are presented in Supplementary Figure 11. Scale bar is 10 μm.

Supplementary Figure 5 Phosphorylation of the CK2 site in Brd4.

(a) Dot blot for phopho-Brd4 antisera using target and control peptides shows specific binding to peptides containing phosphorylated S492. (b) Western blot with phopho-Brd4 antisera in lyates treated with phosphatase or control lysates. Blots are representative of 3 replicates. (c) Quantification of Brd4 expression shows elevated and equivalent Brd4 levels in neurons transfected with GFP and Brd4 with deletions or mutations in the CK2 site (unpaired two-sided t test, for GFP n = 68, for Brd4 n = 61, P = 4.596E-17, t = 9.73, for CK2 deletion n = 46, P = 1.26E-27, t = 14.57 for deletion 492-494 n = 44, P = 1.48E-22, t = 12.37, for S492A n = 54, P = 4.14E-34, t = 17.17, for SSS492ESE n = 51 from 5 biological replicates, P = 7.82E-24, t = 12.73, no significant differences between Brd4 constructs). (d) Example images of EGFP-Brd4 with CK2 site mutations costained with H4K16acetyl. (e) Example graphs showing corresponding changes in fluorescent signal of Brd4 and H4K16acetyl in sections of neuronal nuclei. **, p<0.01. ***, p<0.001. Full-length blots are presented in Supplementary Figure 11. Error bars represent standard error. Scale bar is 5 μm.

Supplementary Figure 6 Brd4 inhibition decreases surface GluA1.

(a) Quantification of mRNA after different times of Jq1 treatment. N = 3 biological replicates for Arc and Fos, N = 8 for Nr4a1, N = 9 for Gria1, N = 4 for Bdnf and N = 5 for Brd4. (b) Quantification of hippocampal neurons treated with Jq1 for 24 hours and stained for surface GluA1 (unpaired two-sided t test, for control n = 13 neurons and for Jq1 n = 12 neurons from 2 biological replicates, P = 0.0015, t = 3.56). (c, d) Images (c) and quantification (d) of surface GluA1 staining 5 days after transfection with different siRNAs targeted against Brd4 (unpaired two-sided t test, for control siRNA n = 14 neurons, for Brd4 siRNA1 n = 13 neurons, and for Brd4 siRNA2 n = 14 neurons from 2 biological replicates, for control vs siRNA1 P = 0.0114, t = 2.61, for control vs siRNA2 P = 0.0078, t = 2.76). (e) Quantification of spine number in neurons treated with Jq1 for 24 hours (unpaired two-sided t test, n = 68 neurons for control for Jq1 and n = 66 from 2 biological replicates). (f) Quantification of staining of surface GluA1 in neurons with long or short forms of Brd4 (unpaired two-sided t test, for control n = 15 neurons, for long Brd4 n = 17, and for short Brd4 n = 18 from 2 biological replicates, for control vs long Brd4 P = 0.0161, t = 2.55, for long vs short Brd4 P = 0.0308, t = 2.26). (g) ChIP for Brd4 at the gria1 promoter region. N = 7 biological replicates for control conditions, 3 biological replicates for Jq1 conditions. (h) ChIPs for histone acetylation at the gria1 promoter region. N = 2 biological replicates. *, p<0.05. ***, p<0.001. a.u. arbitrary units. n.s., non-significant. Scale bar is 10 μm.

Supplementary Figure 7 Jq1 affects mouse behavior.

(a) Weight change in mice treated daily with Jq1 or vehicle. (b) Zone preference ratio in an open field in mice treated with vehicle or with Jq1 for 1 or 3 weeks. (c) Total distance traveled during 1 hour of an open field in mice treated with vehicle or with Jq1 for 1 or 3 weeks. (d) Time spent moving during 1 hour of an open field in mice treated with vehicle or with Jq1 for 1 or 3 weeks. (e) Distance traveled in the novel object box during habituation. (f-i) Time spent exploring objects for memory tests during initial exposure (f) or during testing (g) for memory tests or for learning tests during initial exposure (h) or during testing (i). (j) Discrimination index of time spent with a novel vs familiar object when mice are tested immediately after initial exposure to objects (for learning tests, n = 10 mice for DMSO and 9 mice for 1 week Jq1. No significant difference between DMSO and Jq1, for one sample t test for DMSO P = 0.00325, t = 4.14, for Jq1 P = 0.00103, t = 5.017). (k) Percent of time spent freezing in response to a tone before training, during training, or 1 day after training in a new context (2-way ANOVA with posthoc t test, P = 2E-16, F = 165.7, df = 130) (l) Percent of time spent freezing before, immediately after, or 1 day after fear conditioning training in the training context or in a novel context. (2-way ANOVA with posthoc t test, P = 2E-16, F = 80.72, df = 110). N = 10 mice for control and 3 week Jq1 and 9 mice for 1 week Jq1 for testing groups. *, p<0.05. ***, p<0.001. n.s., nonsignificant. Error bars represent standard error.

Supplementary Figure 8 Jq1 affects seizure susceptibility.

(a) Quantification of mRNA from cortical tissue from mice after behavioral testing following treatment with DMSO or 1 week of Jq1 (unpaired two-sided t test for dmso n = 9 brains and for Jq1 n = 8, for gria1 P = 0.00517, t value = 3.27, for Nr4a1 P = 0.018, t value = 2.64). (b) Western of GluA1 levels after Jq1 treatment. (c) Data for each level of Racine scoring of seizure induction for mice treated for one week with either DMSO or Jq1 and then given pentylenetetrazol (PTZ) to induce seizures. (d) Percent mortality of male mice after seizure injection by PTZ. (e) Seizure susceptibility score of female mice treated for one week with either DMSO or Jq1 and given pentylenetetrazol (PTZ) to induce seizures. (f) Percent mortality of female mice after seizure injection by PTZ. (g, h) Seizure susceptibility scores during kindling for individual mice treated with DMSO (f) or Jq1 (g). N = 9 mice per group. *, p<0.05. ***, p<0.001. Error bars represent standard error. Full-length blots are presented in Supplementary Figure 11.

Supplementary Figure 9 Model of Brd4 function in neurons.

Neuronal activation by signaling molecules such as BDNF trigger activation of CK2 which in turn phosphorylates Brd4. Brd4 binds to acetylated lysines on histone proteins and activates expression of neuronal genes including IEGs which then affect synaptic proteins. Inhibition of Brd4 by Jq1 affects aspects synaptic function and memory formation.

Supplementary Figure 10 Original western blots for Figure 1 and Supplementary Figures 1 and 2.

Supplementary Figure 11 Original western blots for Figures 3, 4, 5, 6 and Supplementary Figures 4, 5 and 8.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Korb, E., Herre, M., Zucker-Scharff, I. et al. BET protein Brd4 activates transcription in neurons and BET inhibitor Jq1 blocks memory in mice. Nat Neurosci 18, 1464–1473 (2015). https://doi.org/10.1038/nn.4095

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4095

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing